EP0546939A1 - Biodegradable magnetic microclusters and methods for making them - Google Patents

Biodegradable magnetic microclusters and methods for making them Download PDF

Info

Publication number
EP0546939A1
EP0546939A1 EP92403345A EP92403345A EP0546939A1 EP 0546939 A1 EP0546939 A1 EP 0546939A1 EP 92403345 A EP92403345 A EP 92403345A EP 92403345 A EP92403345 A EP 92403345A EP 0546939 A1 EP0546939 A1 EP 0546939A1
Authority
EP
European Patent Office
Prior art keywords
magnetic
particles
polymer
microcluster
metal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP92403345A
Other languages
German (de)
French (fr)
Inventor
Mark S. Chagnon
John R. Ferris
Barry L. Fiore
Michelle J. Carter
Tracy J. Hamilton
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Molecular BioQuest Inc
Original Assignee
Molecular BioQuest Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Molecular BioQuest Inc filed Critical Molecular BioQuest Inc
Publication of EP0546939A1 publication Critical patent/EP0546939A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6941Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a granulate or an agglomerate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/183Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an inorganic material or being composed of an inorganic material entrapping the MRI-active nucleus, e.g. silica core doped with a MRI-active nucleus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1833Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with a small organic molecule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1851Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule
    • A61K49/1854Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule the organic macromolecular compound being obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly(meth)acrylate, polyacrylamide, polyvinylpyrrolidone, polyvinylalcohol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1887Agglomerates, clusters, i.e. more than one (super)(para)magnetic microparticle or nanoparticle are aggregated or entrapped in the same maxtrix
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S428/00Stock material or miscellaneous articles
    • Y10S428/90Magnetic feature
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/25Web or sheet containing structurally defined element or component and including a second component containing structurally defined particles
    • Y10T428/256Heavy metal or aluminum or compound thereof
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/26Web or sheet containing structurally defined element or component, the element or component having a specified physical dimension
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2989Microcapsule with solid core [includes liposome]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2991Coated
    • Y10T428/2998Coated including synthetic resin or polymer
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/24Nuclear magnetic resonance, electron spin resonance or other spin effects or mass spectrometry

Definitions

  • NMR has found increasing use since the early 1970's as a medical diagnostic tool, in particular as an imaging technique.
  • the technique provides high resolution and differentiation of soft tissue without the use of potentially harmful radiation.
  • radiologists believed that with the high contrast achieved in NMR imaging in soft tissues without the use of contrast agents, the use of contrast agents would not be necessary.
  • paramagnetic complexes can be used with advantage to achieve enhanced contrast in NMR imaging thereby extending the diagnostic utility of the technique.
  • the nuclei of many atoms have a property called spin which is associated with a small magnetic moment. In the absence of an external magnetic field, the distribution of the orientations of the magnetic moments is random. In the presence of a static magnetic filed the nuclear magnetic moments process about the field direction and there will be a net alignment in the field.
  • T1 and T2 Two additional values can be determined when the RF pulse is turned off and the nuclear magnetic moments are relaxing or returning to equilibrium orientations and phases. These are T1 and T2, the spin-lattice and spin-spin relaxation times.
  • T1 represents a time characteristic of the return to equilibrium spin distribution, i.e. equilibrium alignment of the nuclear magnetic moments in the static field.
  • T2 represents a time characteristic of the return to random precession phase distribution of the nuclear magnetic moments.
  • the NMR signal that is generated thus contains information on proton density, T1 and T2 and the images that are generated are generally the result of complex computer data reconstruction on the basis of that information.
  • contrast agents in extending the diagnostic utility of NMR imaging is discussed, for example, by R.C. Brasch in Radiology 147 :781 (1983). Although numerous methods of contrast are available, many, such as manipulation of tissue temperature, viscosity or hydration, are clearly not clinically feasible and the most advantageous prior art technique appears to be the use of paramagnetic contrast agents to reduce the spin-lattice relaxation of time T1.
  • a paramagnetic substance is one which contains one or more fundamental particles (electrons, protons or neutrons) with a spin whose effect is not cancelled out by another particle with like spin. These particles create a small magnetic field which can interact with neighboring nuclear magnetic dipoles to cause a reorientation of the dipole, i.e. a change in nuclear spin and precession phase.
  • Superparamagnetic particles have also been shown to be effective for the delivery and targeting of drugs directly to an infected organ, tissue or joint. Delivery systems, for example, using magnetic particles 100 Angstroms (A) in diameter encapsulated in albumin microspheres have been demonstrated for delivery of chemotherapeutic agents into Yoshida rat sarcoma. Widder, U.S. Patent 4,345,588 (1982); Senyei et al., U.S. Patent 4,357,259 (1982).
  • the present invention is based on the discovery that, remarkably, when a population of non-magnetic particles of iron metal, magnetic iron oxide or magnetic metal alloy having a diameter of about 70 A or less are linked or encapsulated into a bead structure of about 100 A or greater in diameter, the bead will behave as a superparamagnetic species. If the bead is designed with appropriate chemistry rendering the bead biodegradable, the magnetizable particle will, upon degradation to the unit crystal size, become non-magnetic.
  • Each crystallite is a subdomain (less than 70 A) crystal, or group of crystals, of a transition metal, alloy or metal oxides comprised of trivalent and divalent cations of the same or different transition metals or magnetic metal crystal group.
  • Metals, alloys and oxides which are useful as magnetic core material in the present invention include the metals, alloys, and oxides based on metals which appear in the Periodic Table in Groups 4a and b, 5a and b, 6a and 7a.
  • the present crystallites are preferably between about 0.001 and about 0.007 microns (10 A to 70 A) in diameter and have a surface area of about 25 to 1000 square meters per gram.
  • superparamagnetic iron oxide particles are made by precipitation of divalent (Fe2+) and trivalent (Fe3+) iron salts, for example, FeCl2 and FeCl3, in base and then milled to produce the sub 50 A particles.
  • the ratio of Fe2+ and Fe3+ can be varied without substantial changes in the final product by increasing the amount of Fe2+ while maintaining a constant molar amount of iron.
  • a Fe2+/Fe3+ ratio of about 2:1 to about 4:1 is useful in the present invention; a ratio of about 2:1 Fe2+:Fe3+ is particularly useful.
  • An Fe2+:Fe3+ ratio of 1:1 produces magnetic particles of slightly inferior quality to those resulting from the higher Fe2+/Fe3+ ratios, the particle size is more heterogeneous than that resulting from Fe3+Fe2+ of 2:1 or 4:1.
  • the metal powder is then collected, e.g. by magnetic separation or by filtration, and added to a commercial ball mill as an acetone slurry in a concentration of about 1-25%.
  • the mill is filled about halfway with 1/4'' stainless steel balls and the slurry is milled for a period of time necessary to form the sub-domain crystallites, generally about 3-60 days.
  • the subdomain particle slurry formed is treated as the magnetite described in the previous section.
  • the crystallites can be made by precipitating metal powders using borohydrides and reducing the particle size by milling the resulting precipitate, for example, in a ball mill.
  • the metal powder is precipitated from an aqueous solution of, for example, Fe+2 or Fe+3 salt, with sodium borohydride.
  • the resulting properties of the metal powder are unaffected by the balance of the counter ion or by the iron metal salt selected. Complete precipitation occurs spontaneously upon addition of the borohydride.
  • the magnetic metal powder is then collected by magnetic separation or filtration, washed with water to remove all soluble salts, and then washed in acetone to remove all residual water.
  • divalent transition metal salts such as magnesium, manganese, cobalt, nickel, zinc and copper salts may be substituted for iron salts in the precipitation and milling procedure to yield magnetic metal oxides.
  • divalent cobalt chloride (CoCl2) for FeCl2 in the above procedure produced ferromagnetic metal oxide particles. Ferromagnetic metal oxide particles such as those produced with CoCl2 can be washed in the absence of magnetic fields by employing conventional techniques of centrifugation or filtration between washings to avoid magnetizing the particles.
  • the material was cooled to room temperature and used as a monomer in making the tetravalent titanium organometallic coating in Example 5.
  • the resulting polymer encapsulated magnetic particle clusters were about 100 Angstroms to 2 microns in diameter and had a saturation magnetization of about 2,000 gauss and had no remnant magnetization. The bead could easily be degraded by dissolving it in hot water and the resulting unit crystals had no magnetization.
  • 10 gm of 30A particles were prepared as in Example 3.
  • the particle slurry was mixed into a glass, 25% 100cc solution of carboxymethy cellulose (mw 50,000 daltons) and transferred into a 16oz glass jar filled 25% with 1/4'' ss balls.
  • the suspension was mixed on a ball mill for a period of 2 hours.
  • the slurry was removed from the jar mill and added to a blender filled with 500cc of acetone.
  • the mixture was agitated in the blender at the highest speed for 10 minutes causing the polymer to flocculate onto the magnetic particle's surface.
  • the magnetic beads were collected.

Abstract

Subdomain metal or metal oxide particles covalently coupled to chemically reactive organic moieties and subsequently reacted together to form biodegradable magnetic microclusters are disclosed. The magnetic microcluster can be used as contrast agents in NMR imaging for the production of images suitable for use in diagnosis, for in vivo delivery and targeting of drugs, as in vivo, biodegradable agents for the sequestering of free metal ions in the treatment of metal driven disease.

Description

    Background of the Invention
  • NMR has found increasing use since the early 1970's as a medical diagnostic tool, in particular as an imaging technique. The technique provides high resolution and differentiation of soft tissue without the use of potentially harmful radiation. For several years, radiologists believed that with the high contrast achieved in NMR imaging in soft tissues without the use of contrast agents, the use of contrast agents would not be necessary. However, it has recently been found that paramagnetic complexes can be used with advantage to achieve enhanced contrast in NMR imaging thereby extending the diagnostic utility of the technique.
  • The nuclei of many atoms have a property called spin which is associated with a small magnetic moment. In the absence of an external magnetic field, the distribution of the orientations of the magnetic moments is random. In the presence of a static magnetic filed the nuclear magnetic moments process about the field direction and there will be a net alignment in the field.
  • In NMR imaging, a patient is placed in a static field and a short radio frequency pulse is applied via a coil surrounding the patient. The radio frequency or RF signal is selected for the specific nuclei which are to be resonated. The RF pulse causes the magnetic moments of the nuclei to align with the new field and to process in phase, and on termination of the pulse moments return to the original distribution of alignments with respect to the static field and to a random distribution of procession phases giving off a nuclear magnetic resonance signal which can be picked up by a receiving coil. The NMR signal is generally from ¹H nuclei and represents a proton density of the tissue being studied. R.S. First, NMR In Medicine In The 1980's (1983).
  • Two additional values can be determined when the RF pulse is turned off and the nuclear magnetic moments are relaxing or returning to equilibrium orientations and phases. These are T1 and T2, the spin-lattice and spin-spin relaxation times. T1 represents a time characteristic of the return to equilibrium spin distribution, i.e. equilibrium alignment of the nuclear magnetic moments in the static field. T2 on the other hand represents a time characteristic of the return to random precession phase distribution of the nuclear magnetic moments.
  • The NMR signal that is generated thus contains information on proton density, T1 and T2 and the images that are generated are generally the result of complex computer data reconstruction on the basis of that information.
  • The potential application of contrast agents in extending the diagnostic utility of NMR imaging is discussed, for example, by R.C. Brasch in Radiology 147:781 (1983). Although numerous methods of contrast are available, many, such as manipulation of tissue temperature, viscosity or hydration, are clearly not clinically feasible and the most advantageous prior art technique appears to be the use of paramagnetic contrast agents to reduce the spin-lattice relaxation of time T1.
  • A paramagnetic substance is one which contains one or more fundamental particles (electrons, protons or neutrons) with a spin whose effect is not cancelled out by another particle with like spin. These particles create a small magnetic field which can interact with neighboring nuclear magnetic dipoles to cause a reorientation of the dipole, i.e. a change in nuclear spin and precession phase.
  • Since the magnetic field created by an electron is much greater than that created by a proton or a neutron, in practice only ions, molecules, radicals or complexes, which are paramagnetic due to the presence of one or more unpaired electrons, are used as paramagnetic NMR contrast agents.
  • The contrast effect of paramagnetic ions and complexes is predominantly the result of reduction in T1. However, paramagnetic stable free radicals will also cause some reduction in T2. R.C. Brasch, Radiology, 147:781 (1983). Nevertheless the relative reduction of T1 is greater than that of T2.
  • The use of paramagnetic contrast agents in NMR imaging has been extensively investigated and solutions and colloidal dispersions of such agents have been proposed for oral and paraenteral administration in conjunction with diagnostic imaging.
  • Ferromagnetic materials have also been used as contrast agents because of their ability to decrease T2. Medonca-Dias and Lauterbur, Magn. Res. Med., 3:328 (1986); Olsson et al, Mag Res. Imaging, 4:437 (1986).
    Ferromagnetic materials have high, positive magnetic susceptibilities and maintain their magnetism in the absence of an applied field. The use of ferromagnetic materials as MRI contrast agents are described, for example, in PCT Application NO. WO86/01112 and PCT Application No. WO85/043301.
  • A third class of magnetic materials, termed superparamagnetic materials, have been used as contrast agents. Saini et al., Radiology, 167:211 (1987); Hahn et al., Soc. Mag Res. Med. 4(22):1537 (1986). Like paramagnetic materials, superparamagnetic materials are characterized by an inability to remain magnetic in the absence of an applied magnetic field. Superparamagnetic materials can have magnetic susceptibilities nearly as high as ferromagnetic materials and far higher than paramagnetic materials. Bean and Livingston, J. Appl. Phys., Supp. 1 to Vol. 30, 1205, (1959).
  • Ferromagnetism and superparamagnetism are properties of lattices rather than ions or gases. Iron oxides such as magnetite and gamma ferric oxide exhibit ferromagnetism or superparamagnetism depending on the size of the crystals comprising the material, with larger crystals being ferromagnetic. G. Bate In: Ferromagnetic Materials, Vol. 2, Wohlfarth (ed.) p. 439.
  • As generally used, superparamagnetic and ferromagnetic materials alter the MR image by decreasing T2 resulting in image darkening. When injected, crystals of these magnetic materials accumulate in the targeted organs or tissues and darken the organs or tissues where they have accumulated.
  • Superparamagnetic particles have also been shown to be effective for the delivery and targeting of drugs directly to an infected organ, tissue or joint. Delivery systems, for example, using magnetic particles 100 Angstroms (A) in diameter encapsulated in albumin microspheres have been demonstrated for delivery of chemotherapeutic agents into Yoshida rat sarcoma. Widder, U.S. Patent 4,345,588 (1982); Senyei et al., U.S. Patent 4,357,259 (1982).
  • All of the aforementioned in vivo applications have the marked disadvantage of the lack of particle or cluster biodegradability. Half lives of Fe₃O₄ 100 A particles, for example, are in excess of 8 months when injected into a patient's body.
  • Particles of less than 50 A in diameter will generally clear from a patient after in vivo application very quickly; however, below 50 A in diameter there is no evidence of domain wall support and particles of this size are non-magnetic.
  • Summary of the Invention
  • The present invention relates to biodegradable superparamagnetic microclusters and methods of their preparation. The present microclusters comprise clusters of metal or metal oxide particles that are about 70 A or less in crystallite size and which are non-magnetic in the unclustered state. The non-magnetic unit crystals ("crystallites") are encapsulated or bonded together to form a superparamagnetic cluster of crystallites having a cluster size of from about 100 A to 2 microns in diameter.
  • In one embodiment of the present invention, the individual non-magnetic crystallites are coated with monomers functionalized to participate in subsequent crosslinking reactions. Accordingly, the monomers are adsorbed or covalently bound to the crystallites, and the crystallites are covalently linked by crosslinking between the coated crystallites, thereby forming magnetic microclusters. For physiological applications, the crosslinks are hydrolyzable bonds which hydrolyze in the physiological environment. In another embodiment, the crystallites can be coated directly with polymer coatings, which encapsulate the crystallites into magnetic microclusters, wherein the microcluster is conveniently degraded by simply dissolving it in a solvent suitable for the given polymer.
  • The magnetic clusters are biodegradable to the unit crystallites and become non-magnetic upon biodegradation. The magnetically responsive microclusters of this invention overcome problems associated with size, surface area, biodegradation,and magnetic character of previously developed magnetic particles. The present microclusters are useful in clinical applications, such as contrast agents for nuclear magnetic resonance imaging.
  • Detailed Description of the Invention
  • The present invention is based on the discovery that, remarkably, when a population of non-magnetic particles of iron metal, magnetic iron oxide or magnetic metal alloy having a diameter of about 70 A or less are linked or encapsulated into a bead structure of about 100 A or greater in diameter, the bead will behave as a superparamagnetic species. If the bead is designed with appropriate chemistry rendering the bead biodegradable, the magnetizable particle will, upon degradation to the unit crystal size, become non-magnetic.
  • The present magnetic microclusters are clusters of particles comprising a core of metal, metal alloy or metal oxide. These 70 A or less particles are referred to herein as "crystallites". The individual crystallites can be coated with a functionalized organo-metallic monomer which is adsorbed onto or covalently bound to the crystallites thereby forming an organo-metallic polymer coating. The functional or reactive terminal groups on the organometallic polymer coating are then reacted together via chemical reactions, e.g. covalent crosslinking, formation of coordination complexes or bioaffinity coupling to form magnetizable microclusters. These magnetizable clusters have a mean cluster size of about 100 A to about 2 microns in diameter and are referred to hereinafter as "microclusters".
  • Each crystallite is a subdomain (less than 70 A) crystal, or group of crystals, of a transition metal, alloy or metal oxides comprised of trivalent and divalent cations of the same or different transition metals or magnetic metal crystal group. Metals, alloys and oxides which are useful as magnetic core material in the present invention include the metals, alloys, and oxides based on metals which appear in the Periodic Table in Groups 4a and b, 5a and b, 6a and 7a. These include, for example, divalent transition metals, such as iron, magnesium, manganese, cobalt, nickel, zinc and copper, alloys of these metals such as iron alloys or oxides (e.g., iron magnesium oxide, iron manganese oxide, iron cobalt oxide, iron nickel oxide, iron zinc oxide and iron copper oxide), cobalt ferrite, samarium cobalt, barium ferrite, and aluminum-nickel-cobalt and metal oxides including magnetite (Fe₃O₄), hematite (Fe₂O₃) and chromium dioxide (CrO₂). By way of illustration, the crystallite may be comprised of crystals of iron or iron oxide, or may consist of a single crystal of an iron oxide or metal alloy.
  • The present crystallites are preferably between about 0.001 and about 0.007 microns (10 A to 70 A) in diameter and have a surface area of about 25 to 1000 square meters per gram.
  • The crystallite particles can be prepared according to the following general procedure: metal salts, or organometallocenes, are precipitated in a base at high temperature and pressure to form fine magnetic metal oxide crystals. The crystals are redispersed, then washed in water and in an electrolyte. Magnetic separation can be used to collect the crystals between washes, as the crystals are generally superparamagnetic at this stage. The crystals are then milled, for example, in a ball mill, under conditions sufficient to form subdomain (less than 50 A) crystallites, which are non-magnetic.
  • In one embodiment of the present invention, superparamagnetic iron oxide particles are made by precipitation of divalent (Fe²⁺) and trivalent (Fe³⁺) iron salts, for example, FeCl₂ and FeCl₃, in base and then milled to produce the sub 50 A particles. The ratio of Fe²⁺ and Fe³⁺ can be varied without substantial changes in the final product by increasing the amount of Fe²⁺ while maintaining a constant molar amount of iron. A Fe²⁺/Fe³⁺ ratio of about 2:1 to about 4:1 is useful in the present invention; a ratio of about 2:1 Fe²⁺:Fe³⁺ is particularly useful. An Fe²⁺:Fe³⁺ ratio of 1:1 produces magnetic particles of slightly inferior quality to those resulting from the higher Fe²⁺/Fe³⁺ ratios, the particle size is more heterogeneous than that resulting from Fe³⁺Fe²⁺ of 2:1 or 4:1.
  • In this embodiment, aqueous solutions of the iron salts are mixed in a base, such as ammonium hydroxide, which results in the formation of a crystalline precipitate of superparamagnetic iron oxide. The precipitate is washed repeatedly with water by magnetically separating and redispersing it until a neutral pH is reached. The precipitate is then washed once in an electrolytic solution, e.g. a sodium chloride solution. The electrolyte step is important to insure fineness of the iron oxide crystals. The precipitate is then washed with a solvent (e.g. acetone) to remove all of the water.
  • The metal powder is then collected, e.g. by magnetic separation or by filtration, and added to a commercial ball mill as an acetone slurry in a concentration of about 1-25%. The mill is filled about halfway with 1/4'' stainless steel balls and the slurry is milled for a period of time necessary to form the sub-domain crystallites, generally about 3-60 days. At the completion of the milling period, the subdomain particle slurry formed is treated as the magnetite described in the previous section.
  • In another embodiment of the present invention, the crystallites can be made by precipitating metal powders using borohydrides and reducing the particle size by milling the resulting precipitate, for example, in a ball mill. In this process, the metal powder is precipitated from an aqueous solution of, for example, Fe⁺² or Fe⁺³ salt, with sodium borohydride. The resulting properties of the metal powder are unaffected by the balance of the counter ion or by the iron metal salt selected. Complete precipitation occurs spontaneously upon addition of the borohydride. The magnetic metal powder is then collected by magnetic separation or filtration, washed with water to remove all soluble salts, and then washed in acetone to remove all residual water. The particle is added as an aqueous slurry in a concentration of about 1-25% by weight to a commercial ball mill filled half way with 1/4'' stainless steel balls and milled for about 10-60 days. At the completion of the milling period, a subdomain metal slurry is formed.
  • In yet another embodiment of this invention, subdomain crystallites are grown directly from solution at high temperature and pressure. For example, an aqueous solution of 2:1 Fe⁺²/Fe⁺³ is added to an aqueous solution of ammonium hydroxide at >60°C and >1 atmosphere of pressure. The pressure and temperature are slowly reduced to begin formation of a crystal seed bead. The reactants are incubated at the lower temperature and pressure until the precipitation is complete and the reagents are completely used. The pressure and temperature of the reaction vessel are then reduced to ambient conditions and the particles are collected by filtration, washed 3 times, e.g., with deionized water, to remove excess reactants and 3 times with a solvent, such as acetone, to remove excess water. The subdomain particles thus prepared are non-magnetic.
  • In another embodiment of this invention, the subdomain crystals are grown by the reaction of a metallocene with a base. In one embodiment of this method, ferrocene is combined with iron II hydroxide. Iron II hydroxide is prepared by reacting an aqueous solution on iron II (ferrous) chloride, for example, with ammonium hydroxide to form a gelatinous precipitate of iron II hydroxide ((FeO(OH)). The iron hydroxide is collected by filtration, transferred into a commercial ball mill filed halfway with 1/4'' stainless balls and one-quarter way with water, and the resulting iron hydroxide slurry is milled for a period of 1-30 days.
  • A second ball mill is one-quarter filled with an aqueous slurry of ferrocene (1-25%) and half filled with 1/4'' stainless balls. The ferrocene slurry is milled for a period of 1-90 days to produce ferrocene crystals in the size range desired for the finished iron oxide crystallites.
  • The contents of the two ball mills are then mixed together and milling is continued for about 1 hour to about 10 days to produce the subdomain crystallites. This method is described in detail in co-pending U.S. Application Serial No. 07/565,801 filed August 10, 1990, by M.S. Chagnon et al, the teachings of which are hereby incorporated herein by reference.
  • Other divalent transition metal salts such as magnesium, manganese, cobalt, nickel, zinc and copper salts may be substituted for iron salts in the precipitation and milling procedure to yield magnetic metal oxides. For example, the substitution of divalent cobalt chloride (CoCl₂) for FeCl₂ in the above procedure produced ferromagnetic metal oxide particles.
    Ferromagnetic metal oxide particles such as those produced with CoCl₂ can be washed in the absence of magnetic fields by employing conventional techniques of centrifugation or filtration between washings to avoid magnetizing the particles.
  • The crystallites can be coated with an organo-metallic monomer material capable of adsorptive or covalently bonding to the magnetic core particles. The organometallic monomers also contain an aliphatic moiety and organic functionality to which a wide variety of organic and/or biological molecules can be coupled. Organo-metallic monomers useful for the present coated particles are organic coordinate complexes of selected transition and/or post transition metals which are capable of forming a stable coordination compound which can be adsorbed onto or covalently bound to the magnetic particle. The organometallic monomers must be capable of crosslinking in situ on the particle surface, thereby forming the organometallic polymer coating.
  • Particularly useful organo-metallic compounds are coordinate complexes formed from selected transition metals (e.g., Fe, Ni, Co, Cr, Ti, Zr, Hf, V, Ta, Nb) and/or post-transition metals (e.g. Sn, Sb). Organo-titanium compounds which are useful include, for example, titanium-tetra-isopropoxide, amino-hexyl-titanium-tri-isopropoxide, amino-propyl-titanium-tri-isopropoxide and carboxyl-hexyl-titanium-tri-isopropoxide. Other compounds which are useful include silicon-tetra-isopropoxide and carbon-tetra-isopropoxide. The monomers must be able to be functionalized in a manner that allows the polymer coating formed therefrom to form covalent bonds with bioaffinity or chemical reactants. For this purpose, the monomers can be post-functionalized or derivatized, if necessary, with an aliphatic "spacer arm" which is terminated with an organic functional group capable of coupling with bioaffinity adsorbents or chemically reacting to form covalent cross linkages or forming coordinate complexes. The "spacer arm" is an aliphatic hydrocarbon having from about 3 to about 30 atoms, e.g. carbon, nitrogen and/or oxygen atoms. The purpose of the spacer arm is to provide a non-reactive linker (or spacer) between the organic functional group and the polymer coating. The organic functional group is generally a reactive group such as an amine (NH₂), carboxy group (COOH), cyanate (CN), phosphate (PO₃H), sulfate (SO₃H), thiol (SH), or hydroxyl (OH) group, or a functional ligand such as a catechin.
  • In one embodiment of the present invention, amino-hexyl-titanium-tri-isoproxide is coated onto the magnetic particle of choice, and thermally crosslinked to form an organo-titanium polymer coating having an aliphatic spacer arm (the hexyl moiety) and organic functional group.
  • In one embodiment of the present method, an organo-titanium compound, such as titanium-tetra-isopropoxide which lacks the spacer arm and organic functional group, is functionalized by reaction with an agent such as 1-hydroxy-6-amino hexane, to form the amino-hexyl-titanium-tri-isopropoxide. A method of coating metal or metal oxide particles with an organometallic coating is described in detail in co-pending U.S. Application Serial No. 07/566,169, filed August 10, 1990, by M.S. Chagnon, the teachings of which are hereby incorporated herein by reference. The functionalized particle can then be reacted, coupled, or crosslinked via the reaction method of choice.
  • In a further embodiment of the present invention, the biodegradable magnetic microclusters can be formed by macromolecular encapsulation of the non-magnetic metal or metal oxide particles. More particularly, the particle crystallites are prepared as a particle slurry and are mixed with a solution of polymer for a time sufficient to substantially disperse the polymer within the slurry. The crystallites are then encapsulated by the addition of a solvent which causes the polymer to flocculate and collapse onto their surface. The encapsulated crystallites thereby form a superparamagnetic cluster having a cluster size of from about 100 A to 2 microns in diameter, with a saturation magnetization of about 2,000 gauss, with no remnant magnetization. Particularly useful polymers include poly(vinyl alcohol), hydroxypropyl cellulose, carboxymethylcellulose, poly(vinyl pyrrolidone), polyurethane-polyester block copolymers, polystyrene and poly(vinyl acetate)-poly(vinyl chloride) copolymers. These clusters can then be conveniently degraded, for example, by dissolving in a solvent suitable for a given polymer. At that point, the particles no longer remain encapsulated and the resulting unit crystals have no magnetization.
  • The microclusters formed by crosslinking or bonding between the non-magnetic crystallites, or by encapsulation of said crystallites, are superparamagnetic in character. These superparamagnetic microclusters can be used in a number of in vitro and/or in vivo applications where magnetic particles are used. For example, a bioaffinity adsorbent can be covalently linked to the organometallic coating, on the microcluster, and the microcluster can then be used in in vitro separations. Methods of covalently linking a bioaffinity adsorbent to an organometallic-coated particle are described in detail, for example, in co-pending U.S. Application 07/566,169, filed August 10, 1990 by M.S. Chagnon, the teachings of which are incorporated herein by reference.
  • The present microclusters because of their unique characteristics are particularly useful for in vivo and in vitro applications, specifically magnetic tracers for homogeneous immunoassays. The microclusters are superparamagnetic, that is, they are responsive to an applied magnetic field, but do not exhibit remnant magnetization once the magnetic field has been removed. The microclusters are biodegradable, and once the cluster has degraded into its component crystallites, the crystallites are non-magnetic. The microclusters are therefore well suited for use in in vivo diagnostic localization of cells or tissues recognized by the particular bioaffinity adsorbent coupled to the particle, and also for magnetically directed delivery of therapeutic agents coupled to the particles to pathological sites. The microclusters are particularly useful for use in magnetic resonance imaging.
  • The invention will now be further illustrated by the following examples.
  • EXAMPLES Example 1: Preparation of Subdomain Magnetite Particles by Precipitation and Subsequent Size Reduction by Milling
  • 200 grams (1.58 moles) of ferrous chloride (VWR Scientific) and 325 grams (2.0 moles) of ferric chloride were dissolved in 3 liters of water. 2000 grams of ammonium hydroxide (VWR Scientific) concentrate were added at a rate of 50 ml/minute under constant agitation, during which time the temperature of the solution was kept between 25 and 40 degrees C. After the addition of the ammonium hydroxide was complete, the magnetic particle (Fe₃O₄) aqueous slurry was allowed to cool to room temperature.
  • The particles were then washed with 5 volumes of water, and collected between each wash. On the final wash step the particles were adjusted to an aqueous slurry volume of 25% and added to a commercial ball mill. The mill was filled 1/2 way with 1/4'' stainless steel balls and the slurry was milled for a period of 60 days to reduce the particles to 30A diameter.
  • Example 2: Preparation of Subdomain Metal Particles by Sodium Borohydride Reduction and Size Reduction by Milling
  • 200 gm (1.58 moles) of ferrous chloride was dissolved in 1 liter of water. 500 gm of dry sodium borohydride were added to the solution to form a fine iron powder precipitate. The precipitate was washed with water and collected by filtration. The filtered powder was resuspended in water and re-filtered. The washing procedure was done 4 additional times. On the final suspension, the slurry was adjusted to a concentrate of 20% and milled as described in Example 1 for a period of 75 days to produce particles with a mean diameter of less than 50 A. The resulting particles had no magnetic field response.
  • Example 3: Preparation of Subdomain Magnetite Particles by Reaction of Particulate Ferrocene and Iron (II) Hydroxide
  • A 100 gm slurry containing 20% by weight ferrocene in water was milled in a commercial ball mill as described in Examples 1 and 2 for 60 days.
  • A second slurry was prepared by the following procedure: An aqueous solution containing 20 gm of ferrous sulfate was precipitated using 50 gm of ammonium hydroxide concentrate to form the gelatinous ferrous hydroxide. The gel was filtered and the filtrate washed with 5-100 gm volumes of water. The washed gel was then made into a 20% aqueous slurry and milled as previously described for 30 days.
  • The ferrocene and hydroxide slurries were mixed and milled together for 3 days to form fine Fe₃O₄ crystallites. The crystallites had a mean diameter of 30 A and were non-responsive to a magnetic field.
  • Example 4: Preparation of Amino-Hexyl-Titanium-Tri-Isopropoxide
  • 0.1 moles of titanium-tri-isopropoxide (Tyzor TPT Dupont, Wilmington, DE) and 0.1 moles of 6-amino-1-hexanol were added to a 50 ml beaker and stirred at room temperature for 1 minute to form 0.1 mole of amino-hexyl-titanium-tri-isopropoxide. The reaction mixture was heated to 70°C for 10 minutes to evaporate the isopropyl alcohol formed during the reaction.
  • The material was cooled to room temperature and used as a monomer in making the tetravalent titanium organometallic coating in Example 5.
  • Example 5: Preparation of Amine Functional Organo-titanate Coated Particle
  • Particles were prepared according to the procedures set out in Examples 1, 2 and 3. The particles were washed 5 times with water and 3 times with acetone to remove the water. N, N-dimethyl formamide (DMF) was added to the precipitate in the following ratio: 10 ml of DMF per gram of particle. The mixture was loaded into an Eiger Mill and milled continuously for 10 minutes. The mixture was then transferred to a beaker and heated with stirring for 30 minutes at 100°C. The amine functional organo-titanate prepared in Example 4 was immediately added after preparation with constant stirring to the mixture in a ratio of 1 g dry Fe₃O₄ per 3 g of amine functional organo-titanate.
  • This mixture was then heated with stirring for 20 minutes at 65 degrees C and then passed through the Eiger Mill for two passes. The resulting material was washed five times with water, the coated particles were collected by filtration and the aqueous waste was decanted.
  • Example 6: Preparation of Hydroxy-Hexyl-Titanium-Tri-Isopropoxide
  • 0.1 moles of titanium-tri-isopropoxide (Tyzor TPT DuPont, Wilmington, DE) and 0.1 moles of 6-hydroxy-1-hexanol were added to a 50 ml beaker and stirred at room temperature for 1 minute to form 0.1 mole of hydroxy-hexyl-titanium-tri-isopropoxide. The reaction mixture was heated to 70 degrees C for 10 minutes to evaporate the isopropyl alcohol formed during the reaction.
  • The material was cooled to room temperature and used as a monomer in making the tetravalent titanium organometallic coating in Example 7.
  • Example 7: Preparation of Alcohol-Functional Organo-titanate Coated Particle
  • Particles were prepared according to the procedures set out in Examples 1, 2 and 3. The particles were washed 5 times with water and 3 times with acetone. N, N-dimethyl formamide (DMF) was added to precipitate in the following ratio: 10 ml of DMF per gram of particle. The mixture was loaded into an Eiger Mill and milled continuously for 10 minutes. The mixture was then transferred to a beaker and heated with stirring for 30 minutes at 100 degrees C. The alcohol functional organo-titanate prepared in Example 6 was immediately added after preparation with constant stirring to the mixture in a ratio of 1 g dry Fe₃O₄ per 3 g of amine functional organo-titanate.
  • This mixture was then heated with stirring for 20 minutes at 65 degrees C and then passed through the Eiger Mill for two passes. The resulting material was washed five times with water, the coated particles were collected by filtration and the aqueous waste was decanted.
  • Example 8: Coated Particles of Dihydroxy-Benzene-Hexyl-Titanium-Tri-Isopropoxide
  • 10 grams of amino functional particles prepared in Example 5 were prepared in an aqueous slurry containing 10% by weight particle. 10 grams of 2, 3-dihydroxy-5-benzoic acid were added to the slurry and dissolved. 5 grams of cyclohexyl carbodiiomide were added to form a C6 amide coupled product with a 2,3 dihydroxy-benzene termination.
  • Example 9:
  • An organo-titanium coated particle was prepared exactly as in Example 4 and 5 except that 6-carboxy-1-hexanol was used in place of 6-amino-1-hexanol to form a carboxy terminated organo-titanium coated particle.
  • Example 10: Formation of a Magnetic Cluster
  • 10 grams of 2,3 dihydroxy-benzene terminated particles as prepared in Example 8, and 10 grams of carboxy terminated magnetic particles as prepared in Example 9 were mixed with 5 grams of sodium molybdate. The reaction mixture was stirred for a period of 24 hours. The resulting materials were molybdenum coordinate particle clusters about 1 micron in diameter that had a saturation magnetization of about 2000 gauss and no remnant magnetization. The particles could then be degraded back to the original 30 Angstrom magnetic particle by exposure to pH 6 acid for 24 hours.
  • Example 11: Formation of a Magnetic Cluster
  • 10 grams of hydroxyl terminated particles as prepared in Example 7 and 10 grams of carboxy terminated magnetic particles as prepared in Example 9 were mixed. To the mixture was added 10 grams of 1 Normal HCl. The reaction mixture was heated to 60 degrees C and stirred for a period of 24 hours. The resulting materials were ester linked magnetic particle clusters about 1 micron in diameter that had a saturation magnetization of about 2000 gauss and no remnant magnetization. The particles could then be degraded back to the original 20 Angstrom magnetic particle by exposure to pH 6 acid for 24 hours.
  • Example 12: Formation of a Magnetic Cluster-Polymer Bead
  • 10gm of 30A particles were prepared as in Example 3. The particle slurry was mixed into an aqueous 25% 100cc solution of polyvinyl alcohol (mw 50,000 daltons) and transferred into a 16oz glass jar filled 25% with 1/4'' ss balls. The suspension was mixed on a ball mill for a period of 2 hours. When the mixing was completed, the slurry was removed from the jar mill and added to a blender filled with 500cc of acetone. The mixture was agitated in the blender at the highest speed for 10 minutes causing the polymer to flocculate onto the magnetic particle's surface. The magnetic beads were collected. The resulting polymer encapsulated magnetic particle clusters were about 100 Angstroms to 2 microns in diameter and had a saturation magnetization of about 2,000 gauss and had no remnant magnetization. The bead could easily be degraded by dissolving it in hot water and the resulting unit crystals had no magnetization.
  • Example 13: Formation of a Magnetic Cluster-Polymer Bead
  • 10gm of 30A particles were prepared as in Example 3. The particle slurry was mixed into a glass, 25% 100cc solution of hydroxy propyl cellulose (mw 50,000 daltons) and transferred into a 16oz glass jar filled 25% with 1/4'' ss balls. The suspension was mixed on a ball mill for a period of 2 hours. When the mixing was completed, the slurry was removed from the jar mill and added to a blender filled with 500cc of acetone. The mixture was agitated in the blender at the highest speed for 10 minutes causing the polymer to flocculate onto the magnetic particle's surface. The magnetic beads were collected. The resulting polymer encapsulated magnetic particle clusters were about 100 Angstroms to 2 microns in diameter and had a saturation magnetization of about 2,000 gauss and had no remnant magnetization. The bead could easily be degraded by dissolving it in hot water and the resulting unit crystals had no magnetization.
  • Example 14: Formation of a Magnetic Cluster-Polymer Bead
  • 10 gm of 30A particles were prepared as in Example 3. The particle slurry was mixed into a glass, 25% 100cc solution of carboxymethy cellulose (mw 50,000 daltons) and transferred into a 16oz glass jar filled 25% with 1/4'' ss balls. The suspension was mixed on a ball mill for a period of 2 hours. When the mixing was completed, the slurry was removed from the jar mill and added to a blender filled with 500cc of acetone. The mixture was agitated in the blender at the highest speed for 10 minutes causing the polymer to flocculate onto the magnetic particle's surface. The magnetic beads were collected. The resulting polymer encapsulated magnetic particle clusters were about 100 Angstroms to 2 microns in diameter and had a saturation magnetization of about 2,000 gauss and had no remnant magnetization. The bead could easily be degraded by dissolving it in hot water and the resulting unit crystals had no magnetization.
  • Example 15: Formation of a Magnetic Cluster-Polymer Bead
  • 10gm of 30A particles were prepared as in Example 3. The particle slurry was mixed into a glass, 25% 100cc solution of poly(vinyl pyrrolidone) (mw 50,000 daltons) and transferred into a 16oz glass jar filled 25% with 1/4'' ss balls. The suspension was mixed on a ball mill for a period of 2 hours. When the mixing was completed, the slurry was removed from the jar mill and added to a blender filled with 500cc of acetone. The mixture was agitated in the blender at the highest speed for 10 minutes causing the polymer to flocculate onto the magnetic particle's surface. The magnetic beads were collected. The resulting polymer encapsulated magnetic particle clusters were about 100 Angstroms to 2 microns in diameter and had a saturation magnetization of about 2,000 gauss and had no remnant magnetization. The bead could easily be degraded by dissolving it in hot water and the resulting unit crystals had no magnetization.
  • Example 16: Formation of a Magnetic Cluster-Polymer Bead
  • 10 gms of 30A particles were prepared as in Example 3. The particle slurry was washed 5x with acetone by magnetic filtration of the suspended particles after each successive wash and decanting the supermagnetic liquid. The particle slurry was then washed 3x in cyclohexanone using the same technique as the acetone washing procedure, and diluted to 50cc with cyclohexanone after the final wash.
  • The suspension was added to 100cc of a 20% solution of polyester polyurethane block co-polymer (BF Goodrich Estane 5719) dissolved in cyclohexanone and mixed in a blender.
  • The slurry was then added to 200cc of acetone in a blender as described in Example 12 and mixed at high speed for 5 minutes causing the urethane to flocculate and collapse onto the particle's surface forming beads about 0.5-1 micron in diameter.
  • The resulting beads had a magnetization of about 2,000 gauss and no remnant magnetization.
  • The beads could easily be degraded to unit crystals by contact in organic solvent or by hydrolytic decomposition of the ester bonds in the back bone of the polymer by boiling the beads in water for 24 hours or by autoclaving an aqueous suspension of the beads for 90 minutes.
  • Example 17: Formation of a Magnetic Cluster-Polymer Bead
  • 10 gms of 30A particles were prepared as in Example 3. The particle slurry was washed 5x with acetone by magnetic filtration of the suspended particles after each successive wash and decanting the supermagnetic liquid. The particle slurry was then washed 3x in cyclohexonone using the same technique as the acetone washing procedure, and diluted to 50cc with cyclohexonone after the final wash.
  • The suspension was added to 100cc of a 20% solution of polystyrene dissolved in toluene and mixed in a blender using a laboratory paddle stirrer for this.
  • The slurry was then added to 200cc of acetone in a blender as described in Example 12 and mixed at high speed for 5 minutes causing the polystyrene to flocculate and collapse onto the particle's surface forming beads about 0.5-1 micron in diameter.
  • The resulting beads had a magnetization of about 2,000 gauss and no remnant magnetization. The beads could easily be degraded to unit crystals by contact in organic solvent.
  • Example 18 Formation of a Magnetic Cluster-Polymer Bead
  • 10 gms of 30A particles were prepared as in Example 3. The particle slurry was washed 5x with acetone by magnetic filtration of the suspended particles after each successive wash and decanting the supermagnetic liquid. The particle slurry was then washed 3x in cyclohexanone using the same technique as the acetone washing procedure, and diluted to 50cc with cyclohexaone after the final wash.
  • The suspension was added to 100cc of a 20% solution of polyvinyl acetate) polyvinyl chloride (Union Carbide VAGH) dissolved in cyclohexanone and mixed in a blender using a laboratory paddle stirrer for this.
  • The slurry was then added to 200cc of acetone in a blender as described in Example 12 and mixed at high speed for 5 minutes causing the urethane to flocculate and collapse onto the particle's surface forming beads about 0.5-1 micron in diameter.
  • The resulting beads had a magnetization of about 2,000 gauss and no remnant magnetization. The beads could easily be degraded to unit crystals by contact in organic solvent or by decomposition of the backbone of the polymer by boiling the beads in CMF for 24 hours.
  • Equivalents
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (10)

  1. A biodegradable magnetic microcluster characterized by comprising a cluster of non-magnetic metal or metal oxide particles having a crystallite size of 70 Angstroms or less which are coated with a functionalized polymer wherein the magnetic microcluster is about 100 Angstroms in size and is formed by chemical interaction between the functional groups of the polymer.
  2. The biodegradable magnetic microcluster of claim 1 characterized in that the polymer is an organometallic polymer, and wherein coating preferably is formed by crosslinking between organometallic monomers which comprise coordinate complexes of a transition or post-transition metal, and a functionalized organic ligand, said monomers preferably including amino-hexyl-titanium triisopopoxide, amino-propyl-titanium triisopropoxide, amino-propyl-titanium triisopropoxide and carboxy-hexyl-titanium triisopropoxide, and wherein the functionalized organic ligand preferably is an amine, carboxyl, cyanate, phosphate, sulfate, thiol, hydroxyl, or catechin.
  3. The biodegradable microcluster of claim 2 characterized in that the functionalized organometallic polymer coating is adsorbed onto or covalently bonded to the crystallites and the functional groups are reacted together via covalent bonding or complexation to form magnetizable microclusters.
  4. The biodegradable magnetic microcluster of claim 1 characterized by having a cluster size of from about 100 Angstroms to 2 microns, and wherein the crystallite particle size preferably is from about 10 Angstroms to about 70 Angstroms and preferably has a surface area of from about 25 to 1000 square meters per gram.
  5. The biodegradable magnetic microcluster of claim 1 characterized in that the cluster is superparamagnetic and the individual particles are non-magnetic.
  6. The biodegradable magnetic microcluster of claim 1 characterized in that the metal or metal oxide particles include iron, magnesium, manganese, cobalt, nickel, zinc, copper, iron alloys, cobalt, ferrite, samarium cobalt, barrium ferrite, aluminum-nickel-cobalt, magnetite, hematite and chromium dioxide.
  7. A biodegradable magnetic microcluster characterized by comprising a cluster of metal or metal oxide particles associated with a macromolecular species, said particle having an individual crystal diameter of 70 Angstroms or less characterized in that said particles are non-magnetic in the unclustered state and a sufficient number of particles are encapsulated within said macromolecular species to provide a magnetic microcluster, preferably of the size 100 Angstroms to 2 microns.
  8. The biodegradable magnetic microcluster of claim 7 characterized in that the macromolecular species comprises poly(vinyl alcohol), hydroxypropyl cellulose, carboxymethylcellulose, poly(vinyl pyrrolidone), polyurethane-polyester copolymers, polystyrene and poly(vinyl acetate)-poly(vinyl chloride) copolymers.
  9. A method for the preparation of biodegradable magnetic microclusters characterized by comprising:
       supplying a magnetic core particle comprising a magnetically responsive metal, metal alloy or metal oxide having a crystallite size of 70 Angstroms or less; and
       coating said particles with a functionalized organo-metallic monomer which can be adsorbed onto or covalently bind to said particles and which are capable of crosslinking on the particle surface to form a functionalized organo-metallic polymer coating; and
       reacting together the functionalized organo-metallic polymer coatings to form magnetizable microclusters.
  10. A method for the preparation of biodegradable magnetic microclusters characterized by comprising:
       supplying a magnetic core particle slurry comprising a magnetically responsive metal, metal alloy or metal oxide having a crystallite size of 70 Angstroms or less; and
       mixing the particle slurry with a solution of polymer for a time sufficient to substantially disperse said polymer within said particle slurry; and
       encapsulating the particles with said polymer by addition of a solvent which flocculates and collapses the polymer onto the surface of the magnetic particles to form a magnetic microcluster.
EP92403345A 1991-12-13 1992-12-09 Biodegradable magnetic microclusters and methods for making them Withdrawn EP0546939A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US806478 1991-12-13
US07/806,478 US5225282A (en) 1991-12-13 1991-12-13 Biodegradable magnetic microcluster comprising non-magnetic metal or metal oxide particles coated with a functionalized polymer

Publications (1)

Publication Number Publication Date
EP0546939A1 true EP0546939A1 (en) 1993-06-16

Family

ID=25194119

Family Applications (1)

Application Number Title Priority Date Filing Date
EP92403345A Withdrawn EP0546939A1 (en) 1991-12-13 1992-12-09 Biodegradable magnetic microclusters and methods for making them

Country Status (3)

Country Link
US (2) US5225282A (en)
EP (1) EP0546939A1 (en)
CA (1) CA2084970A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993026019A1 (en) * 1992-06-08 1993-12-23 Molecular Bioquest, Inc. Preparation of controlled size inorganic particles for use in separations, as magnetic molecular switches, and as inorganic liposomes for medical applications
US5389377A (en) * 1989-12-22 1995-02-14 Molecular Bioquest, Inc. Solid care therapeutic compositions and methods for making same
US5441746A (en) * 1989-12-22 1995-08-15 Molecular Bioquest, Inc. Electromagnetic wave absorbing, surface modified magnetic particles for use in medical applications, and their method of production
EP0928611A1 (en) * 1996-06-10 1999-07-14 Nittetsu Mining Co., Ltd. Medical powder
US5935866A (en) * 1989-12-22 1999-08-10 Binax Nh, Inc. Preparation of sub 100 A magnetic particles and magnetic molecular switches
EP1120648A1 (en) * 1998-09-10 2001-08-01 Fuji Photo Film Co., Ltd. Method of detecting thiol-containing compound
WO2009027937A3 (en) * 2007-08-31 2009-09-24 Koninklijke Philips Electronics N. V. Clustered magnetic particles as tracers for magnetic particle imaging

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5948384A (en) * 1990-09-14 1999-09-07 Syngenix Limited Particulate agents
GB9020075D0 (en) 1990-09-14 1990-10-24 Filler Aaron G Contrast agents for magnetic resonance imaging of axonal transport
US6919067B2 (en) * 1991-09-13 2005-07-19 Syngenix Limited Compositions comprising a tissue glue and therapeutic agents
WO1994021240A2 (en) * 1993-03-17 1994-09-29 Silica Gel Ges.M.B.H Superparamagnetic particles, process for producing the same and their use
EP0721311A4 (en) * 1993-09-14 1996-09-25 Univ Toledo Second sphere complexes as relaxation agents for image enhancement in magnetic resonance imaging
AU687093B2 (en) * 1994-09-27 1998-02-19 Nycomed Imaging As Contrast agent
US5616315A (en) * 1994-10-13 1997-04-01 Gillette Canada Inc. Particles including degradable material and anti-microbial agent
US5660817A (en) * 1994-11-09 1997-08-26 Gillette Canada, Inc. Desensitizing teeth with degradable particles
US5520904A (en) * 1995-01-27 1996-05-28 Mallinckrodt Medical, Inc. Calcium/oxyanion-containing particles with a polymerical alkoxy coating for use in medical diagnostic imaging
US5690109A (en) * 1995-06-23 1997-11-25 Govind; Rakesh Method of destructive, noninvasive hyperpyrexia of tissues and organisms utilizing nuclear magnetic resonance
ES2239349T3 (en) 1996-01-10 2005-09-16 Amersham Health As CONTRAST MEDIUM.
GB9600427D0 (en) * 1996-01-10 1996-03-13 Nycomed Imaging As Contrast media
US5855868A (en) * 1996-04-01 1999-01-05 Nycomed Imaging As Method of T1 -weighted resonance imaging of RES organs
PT915738E (en) * 1996-08-05 2002-07-31 Schering Ag METHOD FOR PREPARING MEDIA OF CONTRAST FOR TOMOGRAPHY OF MAGNETIC RESONANCE
US6083484A (en) 1996-10-17 2000-07-04 Molecular Biosystems, Inc. Microparticles stabilized by polynuclear chromium complexes and their use as ultrasound contrast agents
DK1314400T3 (en) * 1996-12-31 2007-10-15 Altea Therapeutics Corp Microporation of tissues for delivery of bioactive agents
ES2260809T3 (en) * 1997-07-01 2006-11-01 Vlp Watertown Limited Partnership METHOD FOR THE DETERMINATION OF THE SEX OF A MAMMARY PROGENIE.
US6294152B1 (en) 1999-01-11 2001-09-25 The University Of Toledo Iron(III) complexes as contrast agents for image enhancement in magnetic resonance imaging
US7169618B2 (en) * 2000-06-28 2007-01-30 Skold Technology Magnetic particles and methods of producing coated magnetic particles
US7083642B2 (en) * 2000-12-22 2006-08-01 Avantec Vascular Corporation Delivery of therapeutic capable agents
US6939375B2 (en) 2000-12-22 2005-09-06 Avantac Vascular Corporation Apparatus and methods for controlled substance delivery from implanted prostheses
US20050203612A1 (en) * 2000-12-22 2005-09-15 Avantec Vascular Corporation Devices delivering therapeutic agents and methods regarding the same
US20050125054A1 (en) * 2000-12-22 2005-06-09 Avantec Vascular Corporation Devices delivering therapeutic agents and methods regarding the same
US20030033007A1 (en) * 2000-12-22 2003-02-13 Avantec Vascular Corporation Methods and devices for delivery of therapeutic capable agents with variable release profile
US6471980B2 (en) 2000-12-22 2002-10-29 Avantec Vascular Corporation Intravascular delivery of mycophenolic acid
US7077859B2 (en) 2000-12-22 2006-07-18 Avantec Vascular Corporation Apparatus and methods for variably controlled substance delivery from implanted prostheses
US20020082679A1 (en) * 2000-12-22 2002-06-27 Avantec Vascular Corporation Delivery or therapeutic capable agents
US20030050692A1 (en) * 2000-12-22 2003-03-13 Avantec Vascular Corporation Delivery of therapeutic capable agents
WO2003009778A2 (en) * 2001-07-26 2003-02-06 Avantec Vascular Corporation Methods and devices for delivery of therapeutic capable agents with variable release profile
US6921283B2 (en) * 2001-08-27 2005-07-26 Trompeter Electronics, Inc. BNC connector having visual indication
US20030220297A1 (en) 2002-02-01 2003-11-27 Berstein David L. Phosphorus-containing compounds and uses thereof
US20040142384A1 (en) * 2003-01-16 2004-07-22 Cohen Barb Ariel Magnetic separator
KR100572400B1 (en) * 2004-05-11 2006-04-24 재단법인서울대학교산학협력재단 plastic forming goods using vinyl Polymer particles encapsulating semiconductor nanoparticles and manufacturing method thereof
US8287583B2 (en) 2005-01-10 2012-10-16 Taheri Laduca Llc Apparatus and method for deploying an implantable device within the body
US20060258875A1 (en) * 2005-05-10 2006-11-16 Clementine Reyes Methods for manufacturing supported nanocatalysts and methods for using supported nanocatalysts
US8097229B2 (en) 2006-01-17 2012-01-17 Headwaters Technology Innovation, Llc Methods for manufacturing functionalized inorganic oxides and polymers incorporating same
US7632774B2 (en) * 2006-03-30 2009-12-15 Headwaters Technology Innovation, Llc Method for manufacturing supported nanocatalysts having an acid-functionalized support
JP2010517703A (en) 2007-02-09 2010-05-27 タヘリ ラドュカ エルエルシー Vascular graft and method for processing the same
US7557064B2 (en) 2007-08-13 2009-07-07 Headwaters Technology Innovation, Llc Methods for making immobilized aryl-containing ligands
US20090081272A1 (en) * 2007-09-24 2009-03-26 John Clarke Medical devices having a metal particulate composition for controlled diffusion
US8211601B2 (en) * 2009-04-24 2012-07-03 Xerox Corporation Coating for optically suitable and conductive anti-curl back coating layer
CN116396653B (en) * 2023-05-06 2024-02-09 五邑大学 Method for preparing metal-organic polymer coating based on latticed supermolecular monomer

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987002063A1 (en) * 1985-10-04 1987-04-09 Immunicon Corporation Magnetic-polymer particles
WO1991009678A1 (en) * 1989-12-22 1991-07-11 Omni Quest Corporation Organo-metallic coated particles for use in separations
WO1991015243A1 (en) * 1990-04-02 1991-10-17 Cockbain, Julian, Roderick, Michaelson Diagnostic agents
WO1992002940A1 (en) * 1990-08-10 1992-02-20 Omni Quest Corporation Method for producing magnetic microparticles from metallocenes
WO1992004916A2 (en) * 1990-09-14 1992-04-02 St. George's Enterprises Limited Particulate agents

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS56117379A (en) * 1980-02-18 1981-09-14 Mitsubishi Electric Corp Hand-cut editing process
JPS58100224A (en) * 1981-12-10 1983-06-14 Hitachi Maxell Ltd Magnetic recording medium
US4454234A (en) * 1981-12-30 1984-06-12 Czerlinski George H Coated magnetizable microparticles, reversible suspensions thereof, and processes relating thereto
US4731239A (en) * 1983-01-10 1988-03-15 Gordon Robert T Method for enhancing NMR imaging; and diagnostic use
JPH0611008B2 (en) * 1983-11-16 1994-02-09 株式会社東芝 Dust core
JPS60120767A (en) * 1983-12-06 1985-06-28 Toshiba Chem Corp Electrically conductive paint composition
US4675173A (en) * 1985-05-08 1987-06-23 Molecular Biosystems, Inc. Method of magnetic resonance imaging of the liver and spleen
JPS62283419A (en) * 1985-06-26 1987-12-09 ハ−キュルス インコ−ポレ−テッド Magnetic composition
JPS62157322A (en) * 1985-12-28 1987-07-13 Hitachi Maxell Ltd Magnetic recording medium and its production
EP0267282A1 (en) * 1986-04-23 1988-05-18 Battelle Development Corporation Chromatographic apparatus and process
US5213788A (en) * 1988-09-29 1993-05-25 Ranney David F Physically and chemically stabilized polyatomic clusters for magnetic resonance image and spectral enhancement
US5147573A (en) * 1990-11-26 1992-09-15 Omni Quest Corporation Superparamagnetic liquid colloids

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987002063A1 (en) * 1985-10-04 1987-04-09 Immunicon Corporation Magnetic-polymer particles
WO1991009678A1 (en) * 1989-12-22 1991-07-11 Omni Quest Corporation Organo-metallic coated particles for use in separations
WO1991015243A1 (en) * 1990-04-02 1991-10-17 Cockbain, Julian, Roderick, Michaelson Diagnostic agents
WO1992002940A1 (en) * 1990-08-10 1992-02-20 Omni Quest Corporation Method for producing magnetic microparticles from metallocenes
WO1992004916A2 (en) * 1990-09-14 1992-04-02 St. George's Enterprises Limited Particulate agents

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 73, 1970, Columbus, Ohio, US; abstract no. 8659z, *
STN FILE SERVER & FILE MEDLINE NR.92299507 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5389377A (en) * 1989-12-22 1995-02-14 Molecular Bioquest, Inc. Solid care therapeutic compositions and methods for making same
US5441746A (en) * 1989-12-22 1995-08-15 Molecular Bioquest, Inc. Electromagnetic wave absorbing, surface modified magnetic particles for use in medical applications, and their method of production
US5935866A (en) * 1989-12-22 1999-08-10 Binax Nh, Inc. Preparation of sub 100 A magnetic particles and magnetic molecular switches
WO1993026019A1 (en) * 1992-06-08 1993-12-23 Molecular Bioquest, Inc. Preparation of controlled size inorganic particles for use in separations, as magnetic molecular switches, and as inorganic liposomes for medical applications
EP0928611A1 (en) * 1996-06-10 1999-07-14 Nittetsu Mining Co., Ltd. Medical powder
EP0928611A4 (en) * 1996-06-10 1999-12-15 Nittetsu Mining Co Ltd Medical powder
US6162469A (en) * 1996-06-10 2000-12-19 Nittetsu Mining Co., Ltd. Medical powder
EP1120648A1 (en) * 1998-09-10 2001-08-01 Fuji Photo Film Co., Ltd. Method of detecting thiol-containing compound
EP1120648A4 (en) * 1998-09-10 2002-10-16 Fuji Photo Film Co Ltd Method of detecting thiol-containing compound
US6790609B1 (en) 1998-09-10 2004-09-14 Fuji Photo Film Co., Ltd. Thin membrane for detecting thiol-containing compounds
WO2009027937A3 (en) * 2007-08-31 2009-09-24 Koninklijke Philips Electronics N. V. Clustered magnetic particles as tracers for magnetic particle imaging
JP2010537971A (en) * 2007-08-31 2010-12-09 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Collective magnetic powder as a tracer for magnetic powder imaging

Also Published As

Publication number Publication date
CA2084970A1 (en) 1993-06-14
US5382468A (en) 1995-01-17
US5225282A (en) 1993-07-06

Similar Documents

Publication Publication Date Title
US5225282A (en) Biodegradable magnetic microcluster comprising non-magnetic metal or metal oxide particles coated with a functionalized polymer
US5069216A (en) Silanized biodegradable super paramagnetic metal oxides as contrast agents for imaging the gastrointestinal tract
US4951675A (en) Biodegradable superparamagnetic metal oxides as contrast agents for MR imaging
US4827945A (en) Biologically degradable superparamagnetic materials for use in clinical applications
US5219554A (en) Hydrated biodegradable superparamagnetic metal oxides
EP0177545B1 (en) Use of ferromagnetic particles in contrast agents for nmr imaging and contrast agents
US4770183A (en) Biologically degradable superparamagnetic particles for use as nuclear magnetic resonance imaging agents
US9375495B2 (en) Magnetic resonance imaging contrast agents comprising zinc-containing magnetic metal oxide nanoparticles
Thapa et al. Enhanced MRI T 2 relaxivity in contrast-probed anchor-free PEGylated iron oxide nanoparticles
US5071076A (en) Method for producing magnetic microparticles from metallocenes
JP3306810B2 (en) Ultrafine crystalline iron oxide magnetic particles, their production method and use in medical diagnosis and treatment
US5547682A (en) Preparation and use of novel injectable RES avoiding inorganic particles for medical application
US7976825B2 (en) Cancer cell diagnosis by targeting delivery of nanodevices
CN111330023B (en) Magnetic nano composite material and preparation method and application thereof
JPH07500823A (en) Processed apatite particles for medical diagnostic imaging
CN108030933B (en) High-sensitivity bimodal magnetic resonance contrast agent and preparation method thereof
KR101779283B1 (en) Tl-T2 dual-mode magnetic resonance imaging contrast agent
CN102225866B (en) Preparation method for ferrite nano-particles
Najafypour et al. Alternating magnetic field and ultrasound waves as size controlling parameters in preparation of superparamagnetic Fe3O4 nanoparticles
Eguía-Eguía et al. Magnetic domains orientation in (Fe3O4/γ-Fe2O3) nanoparticles coated by Gadolinium-diethylenetriaminepentaacetic acid (Gd3+-DTPA)
KR101080581B1 (en) Iron oxide/Manganese oxide hybrid nanocrystals for simultaneous T1 and T2 contrast enhancements in MRI and preparation thereof
Thomassen et al. Structure activity relationship of magnetic particles as MR contrast agents
Kroll Synthesis and characterization of superparamagnetic iron oxide-alginate hydrogels and fluid
CN116942639A (en) Preparation method of nano particles based on exosomes and metal-organic framework structure
CN102881398A (en) Polyamino acid coated nano-magnetic fluid composite material and preparation method for same

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): DE ES FR GB IT

17P Request for examination filed

Effective date: 19930527

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MOLECULAR BIOQUEST, INC.

17Q First examination report despatched

Effective date: 19951215

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 19960426